Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
FASEB J ; 18(7): 887-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15001555

ABSTRACT

Bradykinin (BK), a vasoactive, proinflammatory nonapeptide, promotes cell adhesion molecule (CAM) expression, leukocyte sequestration, inter-endothelial gap formation, and protein extravasation in postcapillary venules. These effects are mediated by bradykinin-1 (B1R) and-2 (B2R) receptors. We delineated some of the mechanisms by which BK could influence chronic inflammation by altering CAM expression on leukocytes, endothelium, and synovium in joint sections of peptidoglycan-polysaccharide-injected Lewis rats. Blocking B1R results in significantly increased joint inflammation. Immunohistochemistry of the B1R antagonist group revealed increased leukocyte and synovial CD11b and CD54 expression and increased CD11b and CD44 endothelial expression. B2R antagonism decreased leukocyte and synovial CD44 and CD54 and endothelial CD11b expression. Although these findings implicate B2R involvement in the acute phase of inflammation by facilitating leukocyte activation (CD11b), homing (CD44), and transmigration (CD54). Treatment with a B2R antagonist did not affect the disease evolution in this model. In contrast, when both BK receptors are blocked, the aggravation of inflammation by B1R blockade is neutralized and there is no difference from the disease-untreated model. Our findings suggest that B1R and B2R signaling show physiologic antagonism. B1R signaling suggests involvement in down-regulation of leukocyte activation, transmigration, and homing. Further studies are needed to evaluate the B1 receptor agonist's role in this model.


Subject(s)
Arthritis/metabolism , Bradykinin/physiology , Cell Adhesion Molecules/biosynthesis , Endothelium, Vascular/drug effects , Leukocytes/drug effects , Synovial Membrane/drug effects , Animals , Arthritis/chemically induced , Arthritis/genetics , Arthritis, Rheumatoid/metabolism , Bradykinin/analogs & derivatives , Bradykinin/biosynthesis , Bradykinin/genetics , Bradykinin/pharmacology , Bradykinin B1 Receptor Antagonists , Bradykinin B2 Receptor Antagonists , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Cell Adhesion/drug effects , Cell Adhesion Molecules/genetics , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Endothelium, Vascular/pathology , Female , Gene Expression Regulation/drug effects , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , L-Selectin/biosynthesis , L-Selectin/genetics , Leukocytes/metabolism , Male , Oligopeptides/pharmacology , Peptidoglycan/toxicity , Prekallikrein/analysis , Rats , Rats, Inbred Lew , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Specific Pathogen-Free Organisms , Synovial Membrane/blood supply , Synovial Membrane/metabolism , Synovial Membrane/pathology
2.
Inflamm Res ; 50(3): 149-55, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11339503

ABSTRACT

OBJECTIVE AND DESIGN: We studied the ability of bradykinin (BK) receptor antagonists type 1 and 2 (B1-RA, B2-RA) to prevent acute inflammation. MATERIAL: A peptidoglycan-polysaccharide (PG-APS)-induced model of arthritis in the Lewis rat was analyzed. TREATMENT: Four groups of animals were studied for 5 days. Treatment was administered subcutaneously (s.c.) 1 mg/kg every 12 h. Group I received PG-APS and was treated with the B2-RA, CP-0597 (DArg-Arg-Pro-Hyp-Gly-Thi-Ser-DTic-NChg-Arg). Group II received PG-APS and was treated with a combined B1 and B2-RA, B9430 (DArg-Arg-Pro-Hyp-Gly-Igl-Ser-Dlgl-Oic-Arg). Group III received PG-APS and albumin control. Group IV received albumin control. METHODS: Joint diameter, liver weight, hematocrit, white blood count and plasma concentrations of prekallikrein, high molecular weight kininogen, HK and IL-beta were measured. Groups were compared by ANOVA. RESULTS: Acute arthritis and hepatomegaly were attenuated in the B2-RA-treated animals (p<0.05). Weight loss was more pronounced in the B1/B2-RA-treated animals. Anemia induced by PG-APS was prevented by B2-RA and B1/B2-RA treatment (p<0.001). A marked decrease in plasma HK to 64% of normal was found in the disease-untreated animals, which was completely normalized by B2-RA treatment and partially attenuated by the B1/B2-RA (78%). The decrease in plasma prekallikrein levels was prevented by combined B1/B2-RA treatment (p<0.05). Finally, elevated plasma IL-1beta levels were lowered by B1/B2-RA treatment and were below detection limits with the B2-RA treatment. CONCLUSIONS: These results indicate that the systemic inflammation is due in part to BK generation which can be blocked by B2-RA, while inhibiting the B1 receptor prevents an anti-inflammatory response.


Subject(s)
Arthritis/drug therapy , Bradykinin Receptor Antagonists , Inflammation/prevention & control , Oligopeptides/therapeutic use , Peptidoglycan/toxicity , Acute Disease , Animals , Arthritis/chemically induced , Bradykinin/analogs & derivatives , Bradykinin/physiology , Bradykinin/therapeutic use , Female , Interleukin-1/blood , Kallikreins/blood , Neutrophils/physiology , Rats , Rats, Inbred Lew , Receptor, Bradykinin B2
3.
Hematol J ; 2(6): 371-7, 2001.
Article in English | MEDLINE | ID: mdl-11920276

ABSTRACT

INTRODUCTION: Preliminary studies have shown that high molecular mass kininogen (HK) inhibits cathepsin G-induced platelet activation. However, the potential mechanism underlying this inhibitory effect remains to be elucidated. MATERIALS AND METHODS: Suspensions of washed and gel-filtered platelets were used in radioligand binding and aggregation studies. The amidolytic activity of cathepsin G was measured using specific chromogenic substrate. Western blot technique was utilised to explore the potential complex formation between cathepsin G and HK. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis was used to analyse the cleavage products of HK. RESULTS: At a concentration of 1 microM, HK completely blocked cathepsin G-induced platelet shape change and secretion of ATP. HK inhibited cathepsin G-induced platelet aggregation in a concentration-dependent manner with an IC(50) of 0.48 microM. Moreover, HK was found to inhibit binding of (125)I-cathepsin G to gel-filtered platelets. (125)I-cathepsin G forms a complex with HK. The complex formation did not affect the amidolytic activity of cathepsin G. HK was proteolysed upon interaction with cathepsin G. CONCLUSION: Our results show that high molecular mass kininogen down-regulates cathepsin G-induced platelet activation by forming a complex with cathepsin G and thus prevents binding of cathepsin G to platelets. These kininogen-cathepsin G interactions may be potential targets for pharmacological intervention.


Subject(s)
Cathepsins/antagonists & inhibitors , Kininogen, High-Molecular-Weight/pharmacology , Platelet Activation/drug effects , Blood Platelets/metabolism , Cathepsin G , Cathepsins/metabolism , Cathepsins/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Humans , Inhibitory Concentration 50 , Kinetics , Kininogen, High-Molecular-Weight/metabolism , Peptide Fragments/analysis , Protein Binding , Serine Endopeptidases
4.
J Lab Clin Med ; 136(4): 292-302, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11039850

ABSTRACT

The thrombospondin-1 (TSP1) structural requirements within its heparin-binding domain (HBD)(30 kd) or within the other domains of the molecule (450 kd) that interact with neutrophils (PMNs) have not been delineated. Synthetic peptides based on the HBD, a TSP1 proteolytic fragment lacking the HBD, a large C-terminal domain of TSP1 (210 kd), a TSP1 recombinant fragment (rTSP1(784-932)), and a monoclonal antibody directed against the TSP1 type 3 repeats (mAb D4.6) were utilized to map such structural requirements on TSP1. Synthetic peptides containing a heparin-binding motif and encompassing residues F16-G33 or A74-S95 of TSP1 competed quantitatively with iodine 125-labeled TSP1 for binding to heparinagarose beads. However, only F16-G33 was a competitor of TSP1 binding to PMNs, suggesting that the sequence F16-G33 within the HBD plays a role in PMN binding. The interaction site within the 450-kd fragment was further narrowed. A TSP1 -derived proteolytic fragment (210 kd), a recombinant TSP1 fragment (rTSP1(784-932)), and a type 3 repeat anti-TSP1 monoclonal antibody (mAb D4.6) competed for the binding of 125I-labeled TSP1 to PMNs. The N-terminal of rTSP1(784-932) and C-terminal sequence analysis of TSP1-210 kd delineated the structural requirements for the second binding region for PMNs-namely, residues A784-N823.


Subject(s)
Blood Platelets/metabolism , Neutrophils/metabolism , Thrombospondin 1/metabolism , Amino Acid Sequence , Antibodies, Monoclonal , Binding Sites/drug effects , Binding Sites/immunology , Blood Platelets/chemistry , Calcium/metabolism , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Heparin/metabolism , Heparin/pharmacology , Humans , Iodine Radioisotopes , Molecular Sequence Data , Neutrophils/cytology , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Peptide Mapping , Protein Structure, Tertiary , Sensitivity and Specificity , Thrombospondin 1/chemistry , Thrombospondin 1/immunology
5.
Thromb Haemost ; 80(5): 816-21, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9843177

ABSTRACT

During severe sepsis there is dramatic activation of both contact proteases and the coagulation pathway. These processes contribute to the development of shock and disseminated intravascular coagulation (DIC) respectively. The Pittsburgh mutant of antitrypsin (358Met-Arg) is a novel protease inhibitor with activity against both thrombin and the contact proteases and should therefore prove beneficial as a therapeutic agent in the management of septic shock. This hypothesis was supported by an earlier study in a pig model where recombinant antitrypsin Pittsburgh (rAT Pittsburgh) at a concentration of 1 microM alleviated some of the features of shock, but did not improve survival. In order to reduce the lethal effects of E. coli sepsis we postulated that a higher concentration of antitrypsin Pittsburgh would be necessary. To test this hypothesis we used rAT Pittsburgh in a primate model. This was chosen in preference to another species as E. coli sepsis in the primate has been well characterised and closely resembles the changes seen in man. Surprisingly this treatment did not alleviate the features of shock and unexpectedly appeared to exacerbate the associated coagulopathy. We propose two possible mechanisms for this unforeseen outcome. The first results from the broad spectrum of activity of antitrypsin Pittsburgh. As well as inhibiting thrombin and the contact proteases, the Pittsburgh mutant also inhibits activated protein C. Inhibition of the protein C system is known to exacerbate septic shock. Secondly, a significant quantity of inactive antitrypsin Pittsburgh, cleaved at the reactive centre, was detected in the plasma of the treated animals. Proteolytically altered serpins, including antitrypsin. have been shown to enhance the inflammatory process. Therefore the accumulation of cleaved rAT Pittsburgh might be expected to exacerbate septic shock.


Subject(s)
Disseminated Intravascular Coagulation/prevention & control , Escherichia coli Infections/complications , Protease Inhibitors/pharmacokinetics , Shock, Septic/drug therapy , alpha 1-Antitrypsin/pharmacokinetics , Animals , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Dogs/blood , Drug Evaluation, Preclinical , Endopeptidases/metabolism , Female , Humans , Papio/blood , Partial Thromboplastin Time , Protease Inhibitors/therapeutic use , Protease Inhibitors/toxicity , Rabbits/blood , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/toxicity , Shock, Septic/complications , Species Specificity , Swine/blood , Treatment Failure , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/therapeutic use , alpha 1-Antitrypsin/toxicity
7.
Thromb Haemost ; 80(1): 114-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9684796

ABSTRACT

The effects of inhibition of tumor necrosis factor (TNF) on cell and protease activation were evaluated in 18 normal volunteers given endotoxin (4 ng/kg, i.v.) after an infusion of low (10 mg/m2 i.v., n = 6) or high dose (60 mg/m2 i.v., n = 6) recombinant human dimeric TNF receptor protein (TNFR:Fc) or its vehicle (placebo n = 6). Activation of the coagulation system occurred by 2 h in the TNFR:Fc vehicle-placebo group manifested by decreased prekallikrein functional levels and increased levels of prothrombin F1+2 fragments (p < 0.0001). High or low dose TNFR:Fc delayed the fall in prekallikrein functional levels by 1 h and 4 h, respectively (p < 0.0002), but did not inhibit the increase in circulating levels of prothrombin F1+2 fragments. In contrast, endothelium activation, characterized by increased levels of tissue plasminogen activator, plasminogen activator inhibitor-1, and von Willebrand Factor antigen was blunted by both low and high dose TNFR:Fc (p < 0.001). While the endotoxin-associated decrease in platelet number was not altered, platelet-derived beta-thromboglobulin peak levels were blunted and delayed by TNFR:Fc (p < 0.02). Increased levels of neutrophil elastase were attenuated by low and high dose TNFR:Fc (p < 0.001). These results suggest that although TNF is functionally linked to the activation of endothelium, neutrophils, coagulation, and fibrinolysis, alternative pathways are present in vivo that result in activation of the kallikrein-kinin system after endotoxin-induced TNF release. These alternative pathways may limit some of the anti-inflammatory effects of TNFR:Fc.


Subject(s)
Antigens, CD/metabolism , Blood Coagulation/drug effects , Endotoxins/pharmacology , Fibrinolysis/drug effects , Kinins/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/pharmacology , Adult , Analysis of Variance , Female , Humans , Male , Receptors, Tumor Necrosis Factor, Type II , Reference Values
8.
Thromb Haemost ; 79(1): 186-94, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9459346

ABSTRACT

Platelet thrombospondin (TSP1) forms a complex with high (HK) and low (LK) molecular weight kininogens. We isolated a proteolytic fragment from HK and LK heavy chains (12 kDa) recognized by TSP1 with a N-terminal sequence, K244ICVGCPRDIP254. Lys244-Pro254 oxidized to cyclic form prevented binding of 125I-LK to TSP1. This effect was abolished by reduction and alkylation. Oxidized peptide KICVGCPRDIP (100 microM) reversed the known inhibitory effects of LK or HK (1 microM), on thrombin-induced platelet activation, suggesting this peptide forms part of the cell binding site on HK and LK for activated platelets. KICVGCPRDIP completely inhibited the binding of 125I-LK to activated platelets. However, the peptide only partially inhibited binding of 125I-HK to platelets, suggesting an additional binding site on the HK light chain. Fluorescein-labeled KICVGCPRDIP bound directly and specifically to activated platelets. A monoclonal antibody directed to TSP1 partially inhibited the binding of 125I-HK to activated but not inactivated platelets. We conclude residues Lys244-Pro254 on kininogen heavy chain is responsible for binding to thrombospondin on the surface of activated platelets.


Subject(s)
Kininogens/blood , Lysine , Peptide Fragments/blood , Platelet Activation , Proline , Thrombospondins/blood , Amino Acid Sequence , Biotin , Humans , Iloprost/pharmacology , Molecular Sequence Data , Molecular Weight , Platelet Aggregation Inhibitors/pharmacology , Protein Structure, Tertiary , Surface Properties
10.
Dig Dis Sci ; 42(11): 2356-66, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9398817

ABSTRACT

We have shown that the contact (kallikrein-kinin) system is involved in the pathogenesis of experimental enterocolitis. We now investigate activation of the contact and coagulation pathways, platelets, and neutrophils in active and inactive ulcerative colitis patients as compared to normal controls. In active ulcerative colitis patients, a significant decrease of plasma prekallikrein, high molecular weight kininogen, and C1 inhibitor levels was observed as compared with controls, as well as prekallikrein activation on western blots. Significant elevation of prothrombin fragment (F1 + 2), which indicates thrombin generation, and elastase-alpha 1-antitrypsin complexes, reflecting neutrophil activation, were found in patients with active disease. Plasma beta-thromboglobulin, a marker of platelet activation, was elevated in both active and inactive disease and appears to be a feature of ulcerative colitis. Activation of contact and coagulation pathways, as well as neutrophils, may mediate inflammation in the active phase of ulcerative colitis.


Subject(s)
Colitis, Ulcerative/physiopathology , Kallikrein-Kinin System , Neutrophil Activation , Platelet Activation , Adult , Blood Coagulation/physiology , Blotting, Western , Female , Fibrinolysis/physiology , Humans , Male , Prekallikrein/analysis , beta-Thromboglobulin/analysis
11.
Blood ; 90(4): 1508-15, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9269768

ABSTRACT

We and others have shown that both high and low molecular mass kininogens are able to inhibit the thrombin-induced aggregation of gel-filtered platelets, indicating that the locus for inhibition resides in the heavy chain. The inhibitory site is present in domain 3, confined to the C-terminal portion of the region encoded by exon 7 (K270-G292), and the minimal effective sequence is a heptapeptide (L271-A277; Kunapuli et al, J Biol Chem 271:11228, 1996). Kininogens inhibit thrombin binding to platelets and thus inhibit thrombin-induced aggregation. The molecular mechanism by which kininogens inhibit thrombin-induced aggregation of platelets is unknown. Thrombin has previously been shown to bind to two receptors on the platelet surface, glycoprotein (GP) Ib-IX-V complex and the hepta-spanning transmembrane receptor coupled to G protein(s). We now show that, unlike its effect on normal platelets, kininogen (2 micromol/L) did not inhibit the thrombin-induced aggregation of Bernard-Soulier platelets, which lack the GP Ib-IX-V complex, suggesting that kininogen interacts either directly or indirectly with that complex and restricts access by thrombin to this receptor. We further show that both recombinant K270-G292 polypeptide and the synthetic peptide L271-A277 derived from high molecular mass kininogen lower thrombin binding to platelets in a manner similar to monoclonal antibodies to or ligands (von Willebrand factor and echicetin) of GP Ib-IX. The anti-GP Ib-IX-V complex antibodies, TM-60 and SZ 2, can inhibit 125I-high molecular mass kininogen binding to platelets. Conversely, kininogen could block the binding of biotinylated TM-60 or of 125I-SZ 2. Kininogen inhibited the binding of biotinylated thrombin bound to a mouse fibroblast cell line transfected with the GP Ib-IX-V complex. These results indicated that kininogen binds to the GP Ib-IX-V complex modulating thrombin binding to platelets and the consequent platelet aggregation. Kininogen can thus serve as an important regulator of the early stages of platelet stimulation by thrombin.


Subject(s)
Blood Platelets/metabolism , Kininogens/physiology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/metabolism , Animals , Antibodies, Monoclonal/metabolism , Bernard-Soulier Syndrome/blood , Blood Platelets/drug effects , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Kininogens/pharmacology , Mice , Peptide Fragments/pharmacology , Peptides/metabolism , Platelet Aggregation/drug effects , Transfection
12.
Proc Assoc Am Physicians ; 109(1): 10-22, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9010912

ABSTRACT

We have developed models of acute and chronic inflammatory arthritis and enterocolitis using peptidoglycan-polysaccharide injected intraperitoneally or subserosally (intramurally) into the distal ileum and cecum. Acute inflammation occurs in both Buffalo and Lewis rats, characterized by inflammation of the injected areas of the intestine. However, only the genetically susceptible Lewis rat develops chronic synovitis and joint erosion or adhesions and granulomatous enterocolitis. In the Lewis rat but not the Buffalo rat, these changes are accompanied by a decrease in plasma prekallikrein and high-molecular-weight kininogen, reflecting activation of the kallikrein-kinin system. Pretreatment with a specific plasma kallikrein inhibitor modulates the acute and chronic arthritis. The same inhibitor partially abrogates the acute changes characteristic of enterocolitis, and preliminary data suggest similar results in the chronic model. The results of these studies indicate that the kallikrein-kinin system plays an important role in arthritis and enterocolitis induced by bacterial products and that kallikrein inhibitors are potential therapeutic agents for inflammatory arthritis and inflammatory bowel disease.


Subject(s)
Arthritis/metabolism , Boron Compounds/pharmacology , Enterocolitis/metabolism , Enzyme Inhibitors/pharmacology , Kallikreins/antagonists & inhibitors , Kinins/metabolism , Oligopeptides/pharmacology , Acute-Phase Proteins/metabolism , Animals , Arthritis/chemically induced , Arthritis/pathology , Disease Models, Animal , Enterocolitis/pathology , Female , Humans , Inflammation , Joints/pathology , Kininogens/metabolism , Rats , Rats, Inbred BUF , Rats, Inbred Lew
13.
Eur J Biochem ; 250(2): 532-8, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9428707

ABSTRACT

We have previously demonstrated a low-affinity (0.8 microM, non-covalent complex formation between high-molecular-mass kininogen (HK) and plasminogen (Plg) which prevented Plg interaction with glioma and endothelial cells. We have now extended our previous observations by exploring the potential complex formation between Plg and low-molecular-mass kininogen (LK) and between LK and HK with Plg cleaved with human neutrophil elastase (HNE). Plg cleavage by HNE (PlgHNE) yielded kringles 1-3, kringle 4 and mini-plasminogen. PlgHNE was subjected to SDS/PAGE under non-reducing conditions, followed by western blotting, and incubated with either 125I-HK or 125I-LK. Autoradiograms revealed that 125I-HK bound to miniplasminogen and to kringles 1-3 but not to kringle 4 and the presence of 10 mM 6-aminohexanoic acid (Ahx) disrupted only the interaction with kringles 1-3. In contrast, 125I-LK bound to miniplasminogen but not to kringles 1-3 or 4 and Ahx had no effect at all. The complex formation of either HK (0.67 microM) or LK (3 microM) with Plg (1.5 microM) did not affect its conversion to plasmin by tissue plasminogen activator (t-PA) (10 U/ml) in the presence of a tissue plasminogen stimulator (0.14 microM). However, the rate of conversion of plasminogen to plasmin by t-PA was affected when platelets were added to the reaction mixture. Since HK (0.83 microM) has been shown to inhibit plasmin-induced platelet aggregation, we investigated whether this inhibitory property is found within the heavy chain shared by HK and LK. We found that LK inhibited plasmin-induced platelet aggregation, but a 4-fold molar excess was required when compared to HK. Compared to plasmin, 3-5-fold molar excess of miniplasmin is required to induce platelet aggregation, indicating the important role of kringles 1-3 for plasmin interactions with these cells. These results indicate that HK and LK-mediated inhibition of plasmin-induced platelet aggregation is likely due to complex formation with kringle 5 without interfering with plasmin's active site. We found an additional interaction between HK and kringles 1-3 enhancing the inhibitory effect, presumably by interfering with plasmin's interaction with platelets. This HK and LK-associated modulation of plasmin-induced platelet aggregation may serve as a template to develop synthetic peptides as novel therapeutic agents to prevent some of the plasmin-associated thrombocytopenia seen during thrombolytic therapy.


Subject(s)
Fibrinolysin/pharmacology , Kininogens/pharmacology , Plasminogen/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Blood Platelets/drug effects , Fibrinolysin/metabolism , Humans , Kininogens/metabolism , Molecular Weight , Platelet Aggregation Inhibitors/metabolism
14.
Immunopharmacology ; 33(1-3): 231-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8856155

ABSTRACT

Severe progressive failure of multiple organ systems has emerged during the past three decades as a common cause of death among patients in intensive care units. Sepsis is now better defined as systemic inflammatory response syndrome (SIRS), but its mortality rate has not changed and it continues to be a major health problem. Endotoxin interacts with plasma proteins and contributes to the pathophysiology of SIRS. Information is limited on the effect of endotoxin on human coagulation and fibrinolytic proteins in vivo, as well as on the cell response involved in the cytokine cascade. For this reason we performed quantitative assays to establish the sequence of events that occurs in vivo in the regulation of the contact and fibrinolytic pathways as well as in the cytokine cascade as a response to a single dose administration of endotoxin to normal non-smoking human volunteers.


Subject(s)
Blood Coagulation/drug effects , Cytokines/physiology , Endotoxins/administration & dosage , Endotoxins/toxicity , Fibrinolysis/drug effects , Blood Coagulation/physiology , Fibrinolysis/physiology , Humans , Injections, Intravenous , Kallikrein-Kinin System/drug effects , Kallikrein-Kinin System/physiology , Multiple Organ Failure/etiology , Multiple Organ Failure/physiopathology , Sepsis/etiology , Sepsis/physiopathology
16.
Dig Dis Sci ; 41(5): 912-20, 1996 May.
Article in English | MEDLINE | ID: mdl-8625762

ABSTRACT

A specific plasma kallikrein inhibitor, Bz-Pro-Phe-boroArg (P8720), was used to define the relationship between the kallikrein-kinin (K-K) system and acute intestinal inflammation induced by bacterial peptidoglycan-polysaccharide (PG-APS) in Lewis rats. Group I received human serum albumin (HSA) intramurally in the intestine and was treated with HSA. Group II received PG-APS and was treated with P8720. Group III received PG-APS and was treated with HSA. P8720 attenuated the decrease of high-molecular-weight kininogen and factor XI activity (group II vs group III, P < 0.01). P8720 therapy significantly but modestly decreased acute intestinal inflammation measured by gross gut score (P < 0.01) and more dramatically reduced the tissue myeloperoxidase activity (P < 0.05), a measure of granulocyte recruitment, in group II compared with group III. We conclude that the K-K system is directly involved in the pathogenesis of the acute phase of experimental acute inflammation. A specific inhibitor may modulate inflammatory bowel disease.


Subject(s)
Boron Compounds/therapeutic use , Enzyme Inhibitors/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Oligopeptides/therapeutic use , Acute Disease , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/chemically induced , Kallikrein-Kinin System/drug effects , Peptidoglycan , Rats , Rats, Inbred Lew , Serum Albumin/therapeutic use , Specific Pathogen-Free Organisms , Streptococcus pyogenes
17.
J Biol Chem ; 271(19): 11228-35, 1996 May 10.
Article in English | MEDLINE | ID: mdl-8626672

ABSTRACT

The ability of kininogens to modulate thrombin-induced aggregation of human platelets has been assigned to domain 3 (D3) in the common heavy chain coded for by exons 7, 8, and 9 of kininogen gene. We expressed each of the exons 7, 8, and 9, and various combinations as glutathione S-transferase fusion proteins in Escherichia coli. Each of the exon products 7 (Lys236-Gln292), 9 (Val293-Gly328), and 8 (Gln329-Met357), and their combinations were evaluated for the ability to inhibit thrombin induced platelet aggregation. Only products containing exon 7 inhibited platelet aggregation induced by thrombin with an IC50 of > 20 microM. A deletion mutant of exon 7 product, polypeptide 7A product (Lys236-Lys270) did not block thrombin-induced platelet aggregation, while 7B product (Thr255-Gln292) and 7C product (Leu271-Gln292) inhibited aggregation. These findings indicated that the inhibitory activity is localized to residues Leu271-Gln292. Peptides Phe279-Ile283 and Phe281-Gln292 did not block thrombin, and Asn275-Phe279 had only minimal inhibitory activity. A heptapeptide Leu271-Ala277 inhibited thrombin-induced aggregation of platelets with an IC50 of 65 microM. The effect is specific for the activation of platelets by thrombin but not ADP or collagen. No evidence for a thrombin-kininogen complex was found, and neither HK nor its derivatives directly inhibited thrombin activity. Knowledge of the critical sequence of kininogen should allow design of compounds that can modulate thrombin activation of platelets.


Subject(s)
Kininogens/pharmacology , Peptide Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thrombin/pharmacology , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Base Sequence , Collagen/pharmacology , Exons , Fibrinogen/metabolism , Humans , In Vitro Techniques , Kinetics , Kininogens/biosynthesis , Kininogens/chemistry , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Oligodeoxyribonucleotides , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Plasmids , Protein Conformation , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Sequence Deletion , Structure-Activity Relationship , Thrombin/antagonists & inhibitors
19.
Gastroenterology ; 110(5): 1467-81, 1996 May.
Article in English | MEDLINE | ID: mdl-8613052

ABSTRACT

BACKGROUND & AIMS: Crohn's disease is characterized by unrestrained inflammation with a genetic component. Genetic susceptibility and activation of the kalli-krein-kinin (contact) system were investigated in experimental enterocolitis and extraintestinal inflammation induced by bacterial polymers. METHODS: Kinetics of inflammation in inbred Lewis and Buffalo rats injected subserosally with peptidoglycan-polysaccharide polymers were correlated with in vivo and in vitro activation of the contact system. RESULTS: Lewis rats had a biphasic course of enterocolitis. Acute inflammation peaked 1 day after injection, gradually decreasing until day 14 when intestinal inflammation spontaneously reactivated and persisted for 16 weeks, accompanied by arthritis, granulomatous hepatitis, anemia, and leukocytosis. Self-limited acute enterocolitis in Buffalo rats resolved by 24 days without extraintestinal involvement. Consumption of the precursor proteins prekalli-krein and high-molecular-weight kininogen indicated activation of the plasma contact system in Lewis rats and closely correlated with chronic intestinal inflammation. Contact system activation did not occur in Buffalo rats, even during acute inflammation. In vitro studies showed a decreased rate of kininogen cleavage in Buffalo plasma. CONCLUSIONS: Selective in vivo and in vitro activation of the contact system in susceptible Lewis rats suggests that this pathway is one determinant of genetic susceptibility to granulomatous enterocolitis and systemic complications.


Subject(s)
Enterocolitis/physiopathology , Granuloma/physiopathology , Kallikrein-Kinin System , Acute-Phase Proteins/metabolism , Animals , Arthritis/physiopathology , Disease Susceptibility , Enterocolitis/genetics , Enterocolitis/metabolism , Enzyme Activation , Female , Granuloma/genetics , Granuloma/metabolism , Kallikreins/metabolism , Kininogens/blood , Kininogens/metabolism , Kinins/metabolism , Liver Diseases/physiopathology , Peptidoglycan , Polymers , Polysaccharides , Prekallikrein/metabolism , Rats , Rats, Inbred BUF , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...